
Keywords: actin cytoskeleton, autosomal dominant polycystic kidney disease, cell motility, cell proliferation, cystic fibrosis transmembrane conductance regulator, fluid secretion, heat shock protein 90, metanephric organ culture
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a debilitating renal neoplastic disorder with limited treatment options. It is characterized by the formation of large fluid-filled cysts that develop from kidney tubules through abnormal cell proliferation and cyst-filling fluid secretion driven by cAMP-dependent Cl− secretion. We tested the effectiveness of the indazole carboxylic acid H2-gamendazole (H2-GMZ), a derivative of lonidamine, to inhibit these processes using in vitro and in vivo models of ADPKD. H2-GMZ was effective in rapidly blocking forskolin-induced, Cl−-mediated short-circuit currents in human ADPKD cells, and it significantly inhibited both cAMP- and epidermal growth factor-induced proliferation of ADPKD cells. Western blot analysis of H2-GMZ-treated ADPKD cells showed decreased phosphorylated ERK and decreased hyperphosphorylated retinoblastoma levels. H2-GMZ treatment also decreased ErbB2, Akt, and cyclin-dependent kinase 4, consistent with inhibition of heat shock protein 90, and it decreased levels of the cystic fibrosis transmembrane conductance regulator Cl− channel protein. H2-GMZ-treated ADPKD cultures contained a higher proportion of smaller cells with fewer and smaller lamellipodia and decreased cytoplasmic actin staining, and they were unable to accomplish wound closure even at low H2-GMZ concentrations, consistent with an alteration in the actin cytoskeleton and decreased cell motility. Experiments using mouse metanephric organ cultures showed that H2-GMZ inhibited cAMP-stimulated cyst growth and enlargement. In vivo, H2-GMZ was effective in slowing postnatal cyst formation and kidney enlargement in the Pkd1flox/flox: Pkhd1-Cre mouse model. Thus, H2-GMZ treatment decreases Cl− secretion, cell proliferation, cell motility, and cyst growth. These properties, along with its reported low toxicity, suggest that H2-GMZ might be an attractive candidate for treatment of ADPKD.
NEW & NOTEWORTHY Autosomal dominant polycystic kidney disease (ADPKD) is a renal neoplastic disorder characterized by the formation of large fluid-filled cysts that develop from kidney tubules through abnormal cell proliferation and cyst-filling fluid secretion driven by cAMP-dependent Cl− secretion. This study shows that the lonidamine derivative H2-GMZ inhibits Cl− secretion, cell proliferation, and cyst growth, suggesting that it might have therapeutic value for the treatment of ADPKD.
INTRODUCTION
Autosomal dominant polycystic kidney disease (ADPKD) affects an estimated 1 in 400 − 1,000 people worldwide (1–4). ADPKD is characterized by the progressive growth of large fluid-filled cysts in a number of ductal organs, but predominantly in the kidneys. The steady growth and enlargement of kidney cysts ultimately leads to end-stage renal disease in ∼50% of patients by the age of 50–60 yr and accounts for 6−9% of patients on renal replacement therapy (2, 5, 6). Many years of productive life are lost due to the debilitating complications of polycystic kidney disease (PKD), which include hypertension, hematuria, abdominal pain, and kidney infection (7, 8). Blood pressure control has been partially effective in the clinical management of the disease (8, 9).
In ADPKD, renal cyst growth requires two mechanisms: proliferation of cyst-lining epithelial cells and secretion of fluid into the cyst lumen (10–12). The cellular second messenger cAMP plays a vital role in promoting both fluid secretion and cell proliferation in ADPKD (13–17). cAMP acts downstream of G protein-coupled receptors, such as the arginine vasopression V2 receptor, to activate PKA. This leads to phosphorylation and activation of cystic fibrosis transmembrane conductance regulator (CFTR) to promote Cl− secretion followed by fluid secretion into the cyst lumen (16, 18). The activation of PKA by cAMP also elicits a strong MAPK response in ADPKD cells, which are primed to be activated by decreased cytosolic Ca2+ caused by PKD mutation (17, 19, 20). As the understanding of the molecular mechanisms of ADPKD progression has grown in recent years, so has the number of potential therapies (1–3, 21–34), including drugs that target cAMP-dependent fluid secretion and target cell growth and proliferation. One such drug is the arginine vasopression V2 receptor antagonist tolvaptan, which has been approved by the United States Food and Drug Administration for the treatment of adult patients with ADPKD (17, 35–39); however, high cost and side effects, including polyuria, nocturia, thirst, and liver complications, limit its use in some patients. Many other drugs that have entered clinical trials for ADPKD have been repurposed from non-PKD indications (3, 40, 41). While many of these drugs show promise, concerns have been raised regarding their long-term use and safety profiles (42–45). To be an effective therapy for ADPKD, drugs will need to be efficacious and well tolerated over decades, as cyst growth is slow. As such, there is a need to develop drugs for PKD that have excellent safety profiles and can target both fluid secretion and cell proliferation to successfully reduce cyst growth over a lifetime while preserving normal renal function.
Indazole-carboxylic acids are a class of drugs with the potential to target the cellular machinery implicated in the pathogenesis of PKD. Lonidamine (LND; Supplemental Fig. S1) is best known for its antispermatogenic and anticancer properties (46, 47). LND has been shown to inhibit cAMP-induced CFTR-mediated anion secretion (48, 49) as an open channel inhibitor of CFTR (50), making it and this class of drugs potential candidates for inhibiting cyst-filling fluid secretion in PKD. LND also affects a number of other cellular processes, including lactate transport and the mitochondrial metabolic pathway for aerobic glycolysis and oxygen consumption (51), inducing apoptosis. More recently, one of the derivatives of LND, gamendazole (GMZ), which was developed at the University of Kansas, was shown to inhibit the heat shock protein (Hsp)90 pathway, although the mechanism is still a matter of investigation (52–54), and to decrease the levels of the proteins Her2 (ErbB2) and Akt in MCF-7 cells (52). In addition, GMZ was also found to be associated with the eukaryotic elongation factor eukaryotic translation elongation factor 1α1 (eEF1A1). GMZ and another derivative, H2-GMZ (Supplemental Fig. S1), have been under investigation as male contraceptives. They have appeared to be safe in animal studies, and their effects on the testis are reported to be reversible (55).
H2-GMZ has been shown (using proteolytic fingerprinting and affinity chromatography) to modulate Hsp90 function by a mechanism similar to the natural product celastrol (53). The molecular chaperone Hsp90 has proven to be an important drug target for the treatment of various neoplastic disorders. Hsp90 is a ubiquitous protein, abundant in the cell and required for normal function (56, 57). Hsp90 promotes the maturation and proper folding of more than a hundred substrate or “client” proteins in the cell. Hsp90 inhibitors are of growing therapeutic interest because client proteins are frequently mutated, overexpressed, or functionally active in cancer, thus making these cells more dependent on a finite supply of Hsp90. Various classes of Hsp90 inhibitors currently being tested as anticancer agents have been reviewed extensively (58–63). These agents bind distinct Hsp90 domains or cochaperones such as cell division cycle 37 (cdc37), inhibit their function and cause proteasomal degradation of their client proteins, including numerous protein kinases, transcription factors, and cell surface receptors, leading to decreased cell proliferation. Many Hsp90 client proteins have been implicated in the pathogenesis of PKD (Supplemental Fig. S2), including but not limited to ErbB2 (64, 65), CFTR (66, 67), Akt (68, 69), cyclin-dependent kinases (Cdks) (70), and proteins of the MEK–ERK pathway (71–74). The Hsp90 inhibitors STA-2842 and STA-9090 (ganetespib) have been shown to reduce cyst size and disease progression in mouse models of PKD (75–77). However, the role of the Hsp90 chaperone complex in human ADPKD cells has not been investigated.
Ultimately, we are interested in developing H2-GMZ and its derivatives as a therapeutic for ADPKD based on the ability of the parent compound, LND, to block CFTR channel activity and inhibit cell proliferation. As such, we tested whether H2-GMZ can target both fluid secretion and cell proliferation in human ADPKD cells. To this end, we investigated the effect of H2-GMZ on CFTR-mediated Cl− secretion in ADPKD cells and whether H2-GMZ can inhibit Hsp90 client proteins and cell motility. We further investigated the potential of H2-GMZ to target cyst formation and enlargement in a mouse metanephric organ culture model and in vivo in a polycystin-1 (Pkd1) conditional mouse. Our results suggest that H2-GMZ could be effective against ADPKD cyst growth.
METHODS
H2-GMZ
H2-GMZ {3-[1-(2,4-dichlorobenzyl)-6-trifluoromethyl-1H-indazol-3-yl]-propionic acid} (JWS-2–72) was initially described in Ref. 52 and United States Patent US8362031B2.
Cell Culture
Primary cultures of human ADPKD cyst-lining epithelial cells and normal human kidney (NHK) cells were obtained from the PKD biomarkers, biomaterials, and cellular models core in the Kansas PKD center. The use of discarded clinical specimens is considered to be not human subjects as researched by regulatory agencies and the institutional board at the University of Kansas Medical Center. The generation and use of primary ADPKD and NHK cells have been previously described in detail (78, 79). Cells were maintained in a 37°C humidified CO2 incubator in DMEM-F-12 medium containing 1% FBS and supplemented with insulin, transferrin, and selenium (ITS). Mouse cortical collecting duct cell line M-1 (80) was maintained in DMEM-F-12 medium containing 5% FBS.
Cl− Secretion Assay
Anion secretion was determined by measuring short-circuit current (ISC) across human ADPKD cells. Briefly, confluent monolayers of ADPKD cells on Snapwell supports were inserted into modified Ussing chambers (Harvard Apparatus), and both surfaces were bathed in a Ringer solution maintained at 37°C and equilibrated in 5% CO2–95% O2. Electrodes were positioned within the chambers, and ISC was measured using two dual-voltage-clamp devices (Warner Instruments). Positive ISC reflects the sum of active transport of cations (e.g., Na+) from the apical to basolateral surface and anions (e.g., Cl−) from the basolateral to apical surface. Benzamil was added to inhibit cation transport via the epithelial Na+ channel (ENaC). Forskolin, a potent cAMP agonist, was added to stimulate Cl− current. ISC was continuously monitored and recorded with LabChart7 (ADInstruments).
Cell Proliferation Assay
ADPKD cell proliferation was measured using the Promega Cell Titer 96 MTT assay kit as previously described (12, 13). Cells were plated at a density of 4 × 103 cells/well in a 96-well cell culture plate in medium containing 1% FBS + ITS. The cells were serum starved for 24 h and treated either with 100 µM cAMP or 25 ng/mL epidermal growth factor (EGF) to stimulate cell proliferation or were placed directly in 5% FBS-containing medium. Increasing concentrations of H2-GMZ or LND were added to the medium to determine their effect on cell proliferation. After 72 h of treatment, MTT assays were performed by adding dye solution to the cells and this reaction was stopped after 4 h. Optical densities measured using a spectrophotometer were considered to be directly proportional to the number of viable cells present.
Mitotic Index
ADPKD cells (5 × 103 cells/well) were plated in four-well chamber slides in medium containing 1% FBS + ITS. After 24 h, the medium was changed to include 5% FBS. The cells were treated with increasing concentrations of H2-GMZ for 16 h, fixed with 4% paraformaldehyde, and permeabilized with 0.2% Triton X-100. The cells were mounted using ProLong Gold antifade reagent containing DAPI (Thermo Fisher Scientific) and observed by fluorescence microscopy. Eight to ten separate fields were examined at ×10 magnification, and the number of mitotic nuclei were counted and expressed as a percentage of the total number of nuclei per field.
Western Blot Analysis
ADPKD cells were seeded on six-well culture plates and treated with 50 μM H2-GMZ for various time points. Cytoplasmic protein extracts were prepared by lysing cells in ice-cold lysis buffer [10 mM Tris·Cl (pH 7.5), 150 mM NaCl, 2 mM EDTA (pH 8.0), 1% Triton X-100, 0.5% Nonidet P-40, 25 mM glycerol 2-phosphate, 1 mM sodium orthovanadate, 1 mM phenylmethylsulphonyl fluoride, and 0.1% (vol/vol) Sigma protease inhibitor cocktail]. Nuclei and other Triton-insoluble components were removed by high-speed centrifugation. Protein concentration was measured using the Pierce BCA assay kit. Total protein (20 μg) was boiled with SDS sample buffer and fractionated on 7.5%, 10%, or 12.5% SDS-PAGE gels. Proteins were transferred to PVDF membranes, and nonspecific binding was blocked with 5% powdered milk in Tris-buffered saline with Tween 20 [TBS-T; 10 mM Tris·Cl (pH 7.5), 150 mM NaCl, and 0.1% Tween 20] for 1 h at room temperature. The blocked membranes were incubated with primary antibodies in 5% powdered milk or 5% BSA (for phospho-proteins) in TBS-T overnight at 4°C. Membranes were then washed three times with TBS-T and incubated with alkaline phosphatase-conjugated secondary antibodies (Sigma) in 5% milk in TBS-T for 30 min at room temperature. Membranes were washed three times with TBS-T, and protein bands were visualized using CDP-star detection reagent (GE Healthcare). Intensity was detected and quantitatively analyzed by the Fluor-S MAX multi-imager system (Bio-Rad). Antibodies used for the Western blot analysis were phosphorylated (p-)ERK, actin (Sigma), ERK1, ERK2, ErbB2, retinoblastoma (Rb), Cdk4 (Santa Cruz Biotechnology), Hsp90, Hsp70, Akt, GAPDH (Cell Signaling), and CFTR (R&D Systems).
Immunoprecipitation
ADPKD cells were plated at a density of 1 × 106 cells/flask in medium containing 5% FBS. Cells were lysed after 24 h in gentle lysis buffer [10 mM Tris·Cl (pH 7.5), 100 mM NaCl, 2 mM EDTA (pH 8.0), 1% Nonidet P-40, and 20 mM sodium molybdate plus protease inhibitors] to prevent dissociation of Hsp90 from client proteins. The lysate (500 µg) was mixed overnight with 0.5 μg ErbB2 antibody and protein A/G beads, washed four times with wash buffer (gentle lysis buffer without NaCl) with protease inhibitors, and boiled in 2× Laemmli sample buffer. The proteins from 20-µg total cell lysate (input) were analyzed alongside the immunoprecipitation by the Western blot analysis.
Immunofluorescence
ADPKD cells (1 × 104 cells/well) were plated in eight-well chamber slides, serum starved for 24 h, and treated for 48 h with 25 ng/mL EGF. Cells were fixed, permeabilized, and blocked with 1% BSA. The cells were then incubated for 1 h with anti-Hsp90 antibody (Cell Signaling), washed with PBS, and incubated for 1 h with FITC-conjugated secondary antibody. Slides were mounted with DAPI-containing mounting medium. Images were captured at ×60. Cultured metanephric kidneys were fixed in 4% paraformaldehyde, and frozen sections were cut. For staining, the sections were subjected to heat retrieval and incubated with anti-proliferating cell nuclear antigen (PCNA) antibody (Sigma) and with anti-mouse Texas red (Jackson Immunoresearch).
Actin Staining
Human ADPKD cells or mouse M-1 cortical collecting duct cells were plated in a four-well chamber slide in 5% FBS-containing medium. After 24 h, the cells were treated with increasing concentrations of H2-GMZ for 16 h. The cells were fixed in 4% paraformaldehyde and permeabilized using 0.2% Triton X-100. The cells were then incubated for 20 min with TRITC-conjugated phalloidin, washed with PBS, and mounted with DAPI-containing mounting medium. Images were captured at ×10 or ×60 using the same exposure settings for treated and untreated cells.
Cell Migration Assay
Human ADPKD cells were plated on a 12-well cell culture plate in 5% FBS + ITS medium and allowed to form a confluent monolayer. The monolayer was wounded by scratching it with a P200 pipette tip, and the medium was changed to include increasing concentrations of H2-GMZ. Images of three different fields per well were captured at ×10 magnification immediately after wounding and at 4, 8, and 24 h postwounding. The area of the wound was measured at each time point using the National Institutes of Health ImageJ program. The percentage of wound closure was calculated by subtracting the area of the wound at any given time point from the 0-h wound area and expressing it as a percentage of the 0-h wound area.
Metanephric Organ Culture
Pkd1m1Bei mice were obtained from the Mutant Mouse Regional Resource Center (University of North Carolina, Chapel Hill, NC) and stabilized onto a C57BL/6 background (>10 backcrosses). This mouse has a point mutation (T to G at 9,248 bp) that causes an M to R substitution affecting the first transmembrane domain of Pkd1 (81). Mouse metanephric kidneys were cultured according to previously described methods (67, 82–84). Metanephroi were dissected from embryonic mice and placed on transparent Falcon 0.4-mm cell culture inserts. DMEM-F-12-defined culture medium (supplemented with 2 mM l-glutamine, 10 mM HEPES, 5 µg/mL insulin, 5 µg/mL transferrin, 2.8 nM selenium, 25 ng/mL prostaglandin E, 32 pg/mL T3, 250 U/mL penicillin, and 250 µg/mL streptomycin) was added under the culture inserts, and organ cultures were maintained in a 37°C humidified CO2 incubator for up to 6 days. 8-bromo-cAMP (8-Br-cAMP; 100 µM) was added to induce cyst formation. This was followed by 1–5 µM H2-GMZ or LND for 3 days. Upon culturing and ∼24 h later (day 1) and each day following (days 2–5), kidneys were photographed using a ×2 or ×4 objective, and the images were acquired and quantified using the analySIS imaging program (Soft Imaging System). Fractional cyst area was calculated as the total tubule dilation area divided by the total kidney area.
Mouse Strains and Treatments
All animal protocols were approved and conducted in accordance with Laboratory Animal Resources of the University of Kansas Medical Center and Institutional Animal Care and Use Committee regulations. Pkd1flox/flox: Pkhd1-Cre mice (73) were generated by breeding Pkd1flox/+: Pkhd1-Cre female mice with Pkd1flox/+: Pkhd1-Cre male mice. H2-GMZ treatment was carried out on Pkd1flox/flox: Pkhd1-Cre mice using daily intraperitoneal injections of 20 mg/kg H2-GMZ from postnatal day (PN)8 to PN18. Both males and females were used for the analysis (see Fig. 7 for details).
Figure 7.
H2-gamendazole (H2-GMZ) treatment decreases cystic burden in vivo in a polycystin-1 (Pkd1) mouse model. H2-GMZ treatment was carried out on Pkd1flox/flox: Pkhd1-Cre mice using daily intraperitoneal injections of 20 mg/kg H2-GMZ from postnatal day 8 (PN8) to postnatal day 18 (PN18). A and B: mice treated with H2-GMZ had a significantly reduced cystic index, smaller kidneys and increased renal parenchyma, decreased kidney weight to body weight (KW/BW), and improved blood urea nitrogen (BUN) levels. Males are indicated by blue circles; females are indicated by red circles. C: survival experiments showed significantly longer survival for H2-GMZ-treated mice (control vs. H2-GMZ: 28.8 ± 5 vs. 67.8 ± 23, P < 0.01). Control males died at the following days: 23, 25, 26, and 33. Control females died at the following days: 27, 27, 30, and 39. H2-GMZ-treated males died at the following days: 63 and 103. H2-GMZ-treated females died at the following days: 48, 49, and 76.
RESULTS
H2-GMZ Inhibits CFTR-Mediated Cl− Secretion by ADPKD Cells
LND has been shown to bind directly to CFTR and inhibit CFTR-dependent transepithelial anion currents across rat epididymal cells (48–50). As such, we were interested in testing the effectiveness of the derivative H2-GMZ as an inhibitor of CFTR-mediated Cl− secreted by ADPKD cells. For this, confluent ADPKD cell monolayers were evaluated in Ussing chambers using ISC assays in the presence of forskolin to stimulate Cl− secretion. ENaC was inhibited using benzamil to eliminate cation absorption. We found that 1 μM H2-GMZ effectively inhibited CFTR-mediated ISC within minutes (Fig. 1). Increasing concentrations of LND (up to 120 μM) did not have an effect, whereas the addition of H2-GMZ after LND treatment, or alone, reduced the anion current significantly (Fig. 1, C and D). Thus, H2-GMZ appears to be much more effective than LND in inhibiting CFTR-mediated Cl− secretion across ADPKD monolayers.
Figure 1.

H2-gamendazole (H2-GMZ) inhibits cystic fibrosis transmembrane conductance regulator (CFTR)-mediated short-circuit current (ISC) in autosomal dominant polycystic kidney disease (ADPKD) monolayers. A and B: apical treatment with H2-GMZ inhibited forskolin (FSK)-induced ISC in ADPKD monolayers. Confluent monolayers of human ADPKD cells were treated apically with benzamil and then FSK to first block the epithelial Na+ channel and then induce CFTR current. Increasing concentrations of H2-GMZ were added, and the change in current was recorded. The graph represents the average of four different monolayers from two ADPKD kidneys. *The effect was statistically significant at P < 0.05 as determined by ANOVA. C and D: effect of lonidamine (LND) on FSK-generated current in ADPKD cells followed by 1 µM H2-GMZ (C) or with 1 µM H2-GMZ alone (D).
H2-GMZ Inhibits Cell Proliferation in ADPKD Cells
ADPKD is a neoplastic disorder in which cyst-lining epithelial cells proliferate and secrete fluid into the lumen causing cyst expansion in the kidney, liver, and other organs. We were interested in examining whether H2-GMZ has an inhibitory effect on the proliferation of ADPKD cells as LND has proven to be an effective antiproliferative, cancer chemotherapeutic agent. An initial study (52) with GMZ showed that it inhibits the proliferation of MCF-7 cells with an IC50 value of ∼100 μM. As shown in Fig. 2A, H2-GMZ inhibited the proliferation of ADPKD cells stimulated either with cAMP or EGF and assayed at 72 h. This inhibition occurred in a dose-dependent manner with an IC50 value of 5–10 μM. In contrast, the IC50 value was close to 50 μM in the presence of 5% FBS (Fig. 2B). To analyze the effect of H2-GMZ on ADPKD cell proliferation at an earlier time point, we stained cells on chamber slides with nuclear DAPI stain after treatment for 16 h with H2-GMZ. H2-GMZ (50 µM) decreased the mitotic index compared with untreated controls, and 100 µM H2-GMZ was even more effective (P < 0.01; Fig. 2C). The results indicate that H2-GMZ effectively arrests the proliferation of ADPKD cystic cells in culture. The IC50 value for inhibition of cell proliferation by LND was similar to that for H2-GMZ (Supplemental Fig. S3).
Figure 2.

Dose-dependent decrease in the proliferation of primary human autosomal dominant polycystic kidney disease (ADPKD) cells treated with H2-gamendazole (H2-GMZ). A and B: dose response of ADPKD cells stimulated with 100 µM cAMP, 25 ng/mL epidermal growth factor (EGF), or 5% FBS and treated with H2-GMZ. After 72 h of treatment, MTT assays were performed. In each group, results are expressed as percentages of the control group (not treated with H2-GMZ). The error bars indicate SEs for n = 6. C: ADPKD cells stimulated with 5% FBS and treated with H2-GMZ showed a significant decrease in the number of mitotic figures. Cells were stained with DAPI to visualize the nuclei. Statistical analysis was done using one-way ANOVA and the Tukey honestly significant difference test for pair-wise comparisons. **P < 0.01. D: inhibition of both phosphorylated (p-)ERK and hyperphosphorylated retinoblastoma (Rb) in H2-GMZ-treated ADPKD cells correlated with the inhibition of cell proliferation. In growing cells, there was a doublet of hyperphosphorylated (top) and hypophosphorylated or nonphosphorylated (bottom) Rb protein. H2-GMZ treatment decreased the levels of hyperphosphorylated Rb (top arrow) consistent with decreased cell proliferation. Cytoplasmic extracts were prepared following 24, 48, or 72 h of 50 µM H2-GMZ treatment in the presence of serum and analyzed by Western blot analysis. The results are representative of three independent experiments.
To determine the mechanism for inhibition of cell proliferation, we tested the effect of H2-GMZ on the levels of p-ERK. ADPKD cells were treated for 24, 48, or 72 h with 50 μM H2-GMZ. Western blot analysis with antibodies against p-ERK and total ERK suggested that H2-GMZ-mediated inhibition of cell proliferation occurs at least in part through inhibition of the MEK–ERK pathway (Fig. 2D), which is consistent with previous work that showed that cAMP-driven ERK activation stimulates ADPKD cell proliferation (13, 14, 19, 71). Hyperphosphorylated Rb levels also decreased with H2-GMZ treatment (Fig. 2D, top band), consistent with inhibition of cell proliferation. As Rb is an important effector of cell-cycle progression, the decrease in hyperphosphorylated Rb as well as p-ERK could contribute to the inhibition of cell proliferation by H2-GMZ.
H2-GMZ Decreases Hsp90 Client Protein Levels in ADPKD Cells
Previous studies had shown that GMZ inhibits the Hsp90 pathway in Sertoli cells (52, 53). As the molecular chaperone Hsp90 has been shown to play an important role in maintaining the abnormal proliferative state in various tumor types, we sought to explore the status of Hsp90 in ADPKD cells and whether H2-GMZ inhibited the Hsp90 pathway in these cells. The presence of Hsp90 in ADPKD cells was checked by immunofluorescence, showing localization in the cytoplasm and nucleus (Fig. 3A). The presence of Hsp90 in the nucleus has been well documented (56, 85). To show that client proteins interact with Hsp90 in ADPKD cells, we conducted pull-down experiments with ErbB2 as a representative Hsp90 client protein that may be relevant to ADPKD progression (64). As shown in Fig. 3B, the endogenous proteins Hsp90 and ErbB2 coimmunoprecipitated from ADPKD cell lysates.
Figure 3.

H2-gamendazole (H2-GMZ) blocks heat shock protein (Hsp)90 function in primary human autosomal dominant polycystic kidney disease (ADPKD) cells. A: ADPKD cells were stained with anti-Hsp90 antibody (green) to show the presence of Hsp90 in the cytoplasm and nucleus. Cells were counterstained with the nuclear stain DAPI (blue). The control well was incubated with FITC-conjugated secondary antibody alone. B: Hsp90 and client protein ErbB2 remain associated in ADPKD cells. ErbB2 was immunoprecipitated from ADPKD cell lysates, and the association with Hsp90 was determined by Western blot analysis. C and D: H2-GMZ treatment decreased Hsp90 client protein levels. ADPKD cells were treated for 24, 48, or 72 h with 50 μM H2-GMZ and analyzed by Western blot analysis. GAPDH and actin served as internal controls. E: H2-GMZ targets Hsp90 client proteins for degradation via the proteasome pathway. ADPKD cells treated with H2-GMZ for 24 h in the presence or absence of MG132, a proteasome inhibitor, were lysed in buffer containing Triton X-100, and the insoluble material was pelleted and analyzed by Western blot analysis. 17-Allylamino-17-demethoxygeldanamycin (17-AAG) was used as a positive control for inhibition of the proteasomal pathway. F: H2-GMZ treatment decreased cystic fibrosis transmembrane conductance regulator (CFTR) levels. All results are representative of three independent experiments with cells from three different ADPKD kidneys. Cdk4, cyclin-dependent kinase 4; IP, immunoprecipitation.
To determine whether H2-GMZ affects the levels of other Hsp90 pathway proteins, ADPKD cells were treated for various times with 50 μM H2-GMZ, and protein levels were determined by Western blot analysis. H2-GMZ treatment decreased the amounts of the known client proteins ErbB2, Akt, and Cdk4, all of which regulate cell proliferation (Fig. 3C). Treatment with H2-GMZ did not increase the levels of Hsp90 or Hsp70 (Fig. 3D). Thus, H2-GMZ appears to affect Hsp90 client proteins without eliciting the heat shock response, a highly desirable outcome for an Hsp90 pathway inhibitor.
H2-GMZ Treatment Leads to Degradation of Hsp90 Client Proteins Through the Proteasome Pathway
Hsp90 inhibitors such as 17-allylamino-17-demethoxygeldanamycin (17-AAG) interfere with the chaperone function of Hsp90 (86), thus targeting client proteins to the ubiquitin-mediated proteasomal degradation pathway. Simultaneous inhibition of Hsp90 function and the proteasome pathway would result in the accumulation of ubiquitinated client proteins in the detergent-insoluble fraction of the cell lysate (87). To check whether H2-GMZ targets Hsp90 client proteins for degradation via the proteasome pathway, we treated primary human ADPKD cells with H2-GMZ for 24 h in the presence or absence of the proteasome inhibitor MG132 and determined the levels of Hsp90 client proteins in the detergent (Triton X-100)-soluble and -insoluble fractions from the cell lysate. Figure 3E shows the accumulation of representative client proteins Akt and Cdk4 in the detergent-insoluble fraction following treatment with H2-GMZ and MG132 (lane 4). 17-AAG served as a positive control (lane 6).
In our experiments, H2-GMZ appeared to inhibit key signaling proteins that require Hsp90 for maturation or activity. Newly synthesized CFTR protein remains dependent on Hsp90 for functional maturation in the endoplasmic reticulum, and the Hsp90 inhibitor geldanamycin leads to accelerated degradation of CFTR protein (88). This prompted us to determine the status of CFTR in ADPKD cells following H2-GMZ treatment. We found that H2-GMZ treatment led to a robust inhibition of CFTR protein levels in a time-dependent manner (Fig. 3F). Thus, H2-GMZ seems to affect CFTR in multiple ways, as a direct channel inhibitor and by causing a decrease in CFTR protein. As such, H2-GMZ should be an efficient inhibitor of Cl−-dependent fluid secretion by ADPKD cyst-lining cells.
H2-GMZ Inhibits ADPKD Cell Migration
ADPKD cells exhibit significantly higher chemotactic migration in response to EGF compared with normal cells (89). Hence, blocking the abnormal motility of ADPKD cells could have a normalizing therapeutic effect. To examine whether H2-GMZ inhibits ADPKD cell motility, we performed a cell migration/wound-healing assay. As little as 10 µM, H2-GMZ significantly inhibited the migration of ADPKD cells in 4 h (Fig. 4, A and B). The inhibition lasted up to 24 h and was dose dependent.
Figure 4.

H2-gamendazole (H2-GMZ) treatment inhibits cell migration. A: confluent monolayers of human autosomal dominant polycystic kidney disease cells were scratched with a P200 pipette tip and then treated with increasing concentrations of H2-GMZ. Wound images were captured at ×10 magnification immediately after wounding and at 4, 8, and 24 h postwounding. B: the percentage of wound closure was calculated by subtracting the area of the wound at any given time point from the 0-h wound area expressed as a percentage of the 0-h wound area. Statistical analysis was done to compare H2-GMZ-treated cells with untreated cells at each time point using one-way ANOVA and the Tukey honestly significant difference test for pair-wise comparisons. #P < 0.05; *P < 0.01. The assay was done in triplicate.
Actin bundling is essential for maintaining cell structure, cytoskeletal dynamics, and cell motility. In the original screen for proteins binding to GMZ, Tash et al. (52) identified the eukaryotic translation elongation factor eEF1A1 as a strong binder to GMZ. eEF1A regulates actin bundling and organization separately from its function in translation (90, 91). As GMZ did not appear to inhibit nucleotide binding by eEF1A1, they hypothesized that GMZ might inhibit actin bundling. To determine whether H2-GMZ can inhibit actin cytoskeletal organization, human ADPKD cells were stained with phalloidin conjugated with the fluorophore TRITC (Fig. 5, A and B). Untreated cells exhibited leading edges (lamellipodia) characteristic of migrating cells, whereas H2-GMZ-treated cells were smaller, without leading edges and with less filamentous actin (F-actin), consistent with an effect on cell motility (Fig. 4B). Similar results were observed in the M-1 mouse collecting duct cell line (Fig. 5B), demonstrating that the effect of H2-GMZ on the cytoskeleton was not unique to ADPKD cells.
Figure 5.

H2-gamendazole (H2-GMZ) treatment affects the actin cytoskeleton. A and B: human autosomal dominant polycystic kidney disease (ADPKD) cells or mouse M-1 cortical collecting duct cells were treated with increasing concentrations of H2-GMZ and stained with phalloidin. Images were captured at ×10 (A) or ×60 (B) using the same exposure settings for treated and untreated cells. Red color shows phalloidin bound to actin filaments in the cell. Blue indicates DAPI-stained nuclei. The assay was done in triplicate.
H2-GMZ Treatment Decreases the Cystic Index in Metanephric Organ Culture
We have previously shown that CFTR channels are functional in embryonic kidneys and are required for cAMP-driven cyst-like tubule expansion (67). As H2-GMZ blocked both cell proliferation and CFTR-mediated fluid secretion in ADPKD cells, we examined the effectiveness of H2-GMZ in inhibiting cAMP-dependent cyst growth and enlargement in mouse embryonic kidneys in organ culture. For this, we used kidneys from Pkd1m1Be +/− or −/− embryos (81). As shown in Fig. 6, 5 μM H2-GMZ significantly inhibited cyst formation.
Figure 6.
H2-gamendazole (H2-GMZ) treatment reduces the cyst burden in cAMP-treated metanephric kidneys. A and B: embryonic day 15.5 (E15.5) mouse kidneys from polycystin-1 (Pkd1) +/− and −/− mice were plated on Transwell membranes and treated with 100 μM cAMP with or without 5 μM H2-GMZ for 4 days. H2-GMZ treatment reduced the cystic index of treated kidneys. Fractional cyst area is the total area of all cysts per kidney represented as a fraction of the total area of the kidney. The number of kidneys of each genotype is shown. *The effect was statistically significant at P < 0.01 as determined by a Student’s t test.
H2-GMZ was also effective when it was added a day after cyst initiation (Supplemental Fig. S4) and was partially effective when it was washed out of the medium following a 24-h treatment period (Supplemental Fig. S5). LND also inhibited cyst growth in Pkd1m1Bei+/− metanephric kidneys (Supplemental Fig. S6) but did not appear to be as effective as the same dose of H2-GMZ. LND at 5 μM did not inhibit cyst growth in Pkd1m1Bei−/− metanephric kidneys (data not shown). H2-GMZ-treated metanephric kidneys stained for PCNA, a marker for cell proliferation, showed a marked decrease in the intensity of PCNA staining compared with controls (Supplemental Fig. S7, A and B). As expected for growing embryonic kidneys, there were many brightly stained vesicles and S-shaped bodies visible in the untreated kidneys whereas there were very few in the H2-GMZ-treated kidneys.
H2-GMZ Treatment Decreases Cyst Progression In Vivo in a Pkd1 Mouse Model
To determine the effects of H2-GMZ in vivo, treatment was carried out on Pkd1flox/flox: Pkhd1-Cre mice using daily intraperitoneal injections of 20 mg/kg H2-GMZ from PN8 to PN18. Mice treated with H2-GMZ had significantly smaller kidneys and increased renal parenchyma (Fig. 7A), reduced cystic index (Fig. 7B), decreased kidney weight-to-body weight ratio, and improved blood urea nitrogen levels (Fig. 7B). Survival experiments (Fig. 7C) showed significantly longer survival for H2-GMZ-treated mice (control vs. H2-GMZ: 28.8 ± 5 vs. 67.8 ± 23, P < 0.01), indicating that H2-GMZ may be effective in prolonging renal function.
DISCUSSION
In this study, we investigated the potential of a new class of drugs for ADPKD. We provide evidence that the novel indazole carboxylic acid derivative H2-GMZ is effective in inhibiting fluid secretion and cell proliferation in human ADPKD cells, mouse metanephric kidneys, and in a rapidly progressive mouse model of PKD. To further understand the mechanism of action of H2-GMZ, we explored its effects on the activities of CFTR, Hsp90, and eEF1A1, three proteins through which H2-GMZ appears to mediate its actions in the cell. Human ADPKD cell monolayers responded to H2-GMZ treatment with decreased CFTR-mediated anion current. We also found that H2-GMZ decreased the levels of a number of Hsp90 client proteins, including CFTR, and targeted them for degradation through the ubiquitin–proteasome pathway. Many of these Hsp90 client proteins are required for cell growth and proliferation (Supplemental Fig. S2). H2-GMZ also inhibited the motility of ADPKD cells and decreased F-actin levels, consistent with an inhibition of the actin-bundling properties of eEF1A1. Furthermore, low micromolar concentrations of H2-GMZ were effective in inhibiting cyst formation in a Pkd1 mutant mouse metanephric organ culture model in which a number of processes mediate the growth and enlargement of cysts in response to cAMP (67). Finally, daily injection of H2-GMZ in a postnatal Pkd1 mouse model showed inhibition of cystogenesis and increased survival indicating that H2-GMZ effectively targets the kidney in vivo.
Hsp90 function is critical for protein folding and function in eukaryotic cells. Protein molecules exist in a crowded state in the cell, which favors intermolecular interactions but risks misfolding and aggregation (56, 57). Hsp90 uses energy derived from ATP to promote the protein conformation required for proper functioning. In various neoplastic disorders, Hsp90 and its client proteins are overexpressed and/or hyperactive (92). This leads to increased signaling and uncontrolled cell growth and division. Targeting Hsp90 to reduce its activity has proven to be beneficial in curbing the growth of many tumors (61, 63, 86, 92–94). The Hsp90 inhibitors STA-2842 and STA-9090 (ganetespib) have been shown to be effective in reducing cyst size and disease progression in mouse models of PKD (75–77). As ADPKD has many of the characteristics of a neoplastic disorder (95), we believe this approach may be useful in treating patients with ADPKD.
In ADPKD, renal epithelial cysts grow and enlarge through a process of abnormal cell proliferation and increased fluid secretion. Each cyst could be considered a fluid-filled tumor (96). Apical Cl− transport by CFTR drives net fluid secretion into the cyst lumen in response to cAMP (12, 16, 97, 98). Cyst-lining cells are poorly differentiated and express elevated proto-oncogenes (99). Overexpression of the oncogenes c-myc, rasT24, and simian virus 40 large T antigen leads to PKD in transgenic mouse models (100–102). A key pathway that is abnormally activated in PKD is the MEK–ERK pathway, as also seen in many types of cancer (13, 19, 71–73, 103, 104). In addition, there is evidence for the activation of EGF family receptors, IGF receptors, and β-catenin in many types of cancer as well as PKD (64, 74, 105, 106). A number of these pathways need Hsp90 for sustained activation. There may be higher than normal levels of certain proteins in ADPKD cells that would use Hsp90 for their continued aberrant function. For example, the EGFR family receptor ErbB2 (Her2), an Hsp90 client protein, is overexpressed in PKD (64, 65). Cyst-lining renal epithelial cells also overexpress CFTR (97, 98, 107). For these reasons, we expect that an inhibitor of the Hsp90 pathway would preferentially target the cystic epithelium rather than the normal renal parenchyma.
H2-GMZ is a novel derivative (Supplemental Fig. S1) of the anticancer drug LND, used to treat various types of cancer (47, 108–110). LND is known to inhibit cellular glucose metabolism, possibly by its effect on mitochondrial hexokinase (110, 111). Drug-resistant breast cancer cells and malignant glioma cells were found to be especially sensitive to LND (110, 111). In addition, LND inhibits CFTR activity in cultured epididymal cells, presumably by direct binding to the pore of the channel (48–50). Although the exact mechanism of antitumor activity of LND is unknown, it is most likely a combination of the activities mentioned above and possibly additional activities. LND was originally described as an antispermatogenic drug (112). In an effort to develop better male contraceptives, various analogs of LND were synthesized, among which GMZ showed the greatest potency and the least side effects (52, 55). As PKD is a chronic condition that might require a lifetime of therapy, a drug with excellent safety and toxicity profiles would be the ideal candidate for PKD therapy. In an animal study, GMZ was very well tolerated (55).
In a study to understand the mechanism of action of GMZ, it was observed that GMZ affects two proteins: the constitutive isoform of Hsp90, Hsp90AB1, and the eukaryotic elongation factor eEF1A1 (52). The mechanism by which GMZ inhibits Hsp90 is currently being investigated. Neither geldanamycin (which binds to the NH2-terminus of Hsp90) nor a novobiocin derivative (which binds the COOH-terminus) could compete with GMZ for Hsp90 binding (52). Therefore, it is possible that GMZ binds to a different site on Hsp90 or that it binds one of the cochaperones in the Hsp90 supercomplex. In fact, a subsequent study has demonstrated that the Hsp90-cdc37 chaperone complex was disrupted following H2-GMZ treatment in the ErbB2-overexpressing cancer cell line SKBr3 (53). Hsp90 inhibition by GMZ was also confirmed by a decrease in client proteins ErbB2 and Akt (52, 53).
In testing the effect of H2-GMZ on Hsp90-mediated activation of key signaling proteins, we found that H2-GMZ treatment effectively decreased ErbB2 and Akt. ErbB2 (Her2) is known to be upregulated in ADPKD cells compared with normal renal epithelial cells, and treatment with ErbB2 inhibitors was found to slow cyst growth in a mouse model of ADPKD (64). ADPKD cyst-lining epithelial cells exhibit misregulation of the phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin pathway (69, 113). Cdk4 is an Hsp90 client protein that phosphorylates Rb protein during the G1 phase of the cell cycle. The H2-GMZ-mediated decreases in Cdk4 and hyperphosphorylated Rb could contribute to the observed decrease in cell proliferation. Consistent with H2-GMZ being an Hsp90 inhibitor, we showed that the unfolded client proteins were directed to the proteasome pathway for degradation.
One of the Hsp90 client proteins, the Cl− channel CFTR, plays an important role in the pathogenesis of ADPKD by mediating fluid secretion into the cyst lumen (67, 114), and CFTR inhibitors have proven effective in reducing cyst growth (66). We found that CFTR-mediated secretion decreases rapidly following treatment with H2-GMZ. Furthermore, CFTR protein levels decrease markedly 24 − 72 h after H2-GMZ treatment. As LND directly binds to and blocks CFTR, it is possible that H2-GMZ acts similarly. Thus, H2-GMZ could be inhibiting CFTR channel activity via direct binding and also indirectly through Hsp90, making this drug an especially attractive candidate for ADPKD therapy.
ADPKD cell motility and migration could be involved in cyst formation and PKD progression as ADPKD cells as a monolayer on solid support migrate faster than normal cells to achieve wound closure (89). The integrity of the actin cytoskeleton is critically important to the ability of cells to migrate. It is known that LND significantly inhibits endothelial cell migration and invasiveness (115). LND treatment caused the disappearance of actin stress fibers and rearrangement of intermediate filaments and microtubules (116). We found that H2-GMZ treatment effectively blocked ADPKD cell migration, perhaps by its effect on eEF1A1 (52). eEF1A1 is essential for the regulation of the actin cytoskeleton (91, 117). Previous experiments have shown that GMZ does not affect the GDP/GTP nucleotide-binding properties of eEF1A1, which are required for it to mediate elongation during translation (52). Therefore, it was postulated that GMZ might inhibit the bundling of F-actin by eEF1A1, a function separate from its role in protein translation (91). Our results indicate that H2-GMZ might disrupt cell migration by inhibiting actin polymerization. It is also possible that H2-GMZ could mediate its effect on cell migration independent of eEF1A1.
A drug like H2-GMZ with low toxicity and multiple cellular targets would be ideal for PKD treatment. One of the drawbacks of traditional Hsp90 inhibitors such as geldanamycin derivatives is the compensatory induction of Hsp90 and Hsp70 (118–120), thus reducing their effectiveness over time. In our study, H2-GMZ did not induce Hsp90 or Hsp70 even after 72 h of treatment. This lack of a heat shock response in treated cells could make the drug more effective in targeting the Hsp90 machinery. Although H2-GMZ was explored as a potential male contraceptive, its antispermatogenic effect may not be considered a drawback for every male patient or for female patients. Depending on the stage of the disease and the age and health of the patient, the long-term advantages may outweigh this potential disadvantage, thus justifying the use of H2-GMZ or other LND derivatives in the treatment of ADPKD, although an effort to develop H2-GMZ analogs without male reproductive effects would be a worthwhile goal of future studies.
Perspectives and Significance
Cyst growth in PKD most likely involves multiple pathogenic processes, including increased cell proliferation and cyst-filling fluid secretion, and the progression of the disease may be further affected by inflammation and the development of fibrosis. Currently, there is only one Federal Drug Administration-approved therapy for PKD, tolvaptan. However, although tolvaptan has been demonstrated to slow cyst growth and kidney enlargement by blocking the vasopression 2 receptor and decreasing renal cAMP, it has a number of drawbacks as a long-term therapy. As such, new targets for therapy and new therapeutic strategies need to be developed. H2-GMZ, because it acts as an Hsp90 inhibitor and has several other direct cellular targets, including CFTR, should be considered in the development of new therapies for ADPKD. Future studies should be designed to determine its full range of biological targets, its effectiveness over the long course of disease progression, and its long-term safety. Ultimately, the ideal treatment for PKD may be a combination of therapies in low tolerable doses that hit multiple critical targets including both cell proliferation and fluid secretion to slow cyst growth and enlargement and to preserve kidney function over the long term.
SUPPLEMENTAL DATA
Supplemental Figs. S1−S7: https://doi.org/10.6084/m9.figshare.20371803.
GRANTS
This work was supported by a University of Kansas Institute for Advancing Medical Innovation Grant and PKD Foundation Grant (to J.P.C.), National Institute of Diabetes and Digestive and Kidney Diseases Grant R01DK081579 (to D.P.W.), and the PKD Biomarkers, Biomaterials, and Cellular Models Core of the Kansas PKD Research and Translation Core Center (Grant U54DK126126).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
S.V.S., J.X.Z., D.P.W., G.I.G., J.S.T., X.L., and J.P.C. conceived and designed research; S.V.S., J.X.Z., B.S.M., G.A.R., and S.R.J. performed experiments; S.V.S., J.X.Z., B.S.M., G.A.R., D.P.W., G.I.G., S.R.J., J.S.T., A.S.L.Y., X.L., and J.P.C. analyzed data; S.V.S., J.X.Z., B.S.M., G.A.R., D.P.W., G.I.G., S.R.J., J.S.T., A.S.L.Y., X.L., and J.P.C., interpreted results of experiments; S.V.S., J.X.Z., B.S.M., D.P.W., X.L., and J.P.C. prepared figures; S.V.S. and J.P.C. drafted manuscript; S.V.S., D.P.W., G.I.G., A.S.L.Y., and J.P.C. edited and revised manuscript; S.V.S., J.X.Z., B.S.M., G.A.R., D.P.W., G.I.G., S.R.J., J.S.T., A.S.L.Y., X.L., and J.P.C. approved final version of manuscript.
REFERENCES
- 1. Yu ASL, El-Ters M, Winklhofer FT. Clinical trials in autosomal dominant polycystic kidney disease. In: Polycystic Kidney Disease, edited by Li X. Brisbane: Codon Publications, 2015, chapt. 6. [PubMed] [Google Scholar]
- 2. Bergmann C, Guay-Woodford LM, Harris PC, Horie S, Peters DJM, Torres VE. Polycystic kidney disease. Nat Rev Dis Primers 4: 50, 2018. doi: 10.1038/s41572-018-0047-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Cornec-Le Gall E, Alam A, Perrone RD. Autosomal dominant polycystic kidney disease. Lancet 393: 919–935, 2019. doi: 10.1016/S0140-6736(18)32782-X. [DOI] [PubMed] [Google Scholar]
- 4. Lanktree MB, Haghighi A, Guiard E, Iliuta IA, Song X, Harris PC, Paterson AD, Pei Y. Prevalence estimates of polycystic kidney and liver disease by population sequencing. J Am Soc Nephrol 29: 2593–2600, 2018. doi: 10.1681/ASN.2018050493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Antignac C, Calvet JP, Germino GG, Grantham JJ, Guay-Woodford LM, Harris PC, Hildebrandt F, Peters DJ, Somlo S, Torres VE, Walz G, Zhou J, Yu AS. The future of polycystic kidney disease research–as seen by the 12 Kaplan awardees. J Am Soc Nephrol 26: 2081–2095, 2015. doi: 10.1681/ASN.2014121192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Grantham JJ, Torres VE. The importance of total kidney volume in evaluating progression of polycystic kidney disease. Nat Rev Nephrol 12: 667–677, 2016. doi: 10.1038/nrneph.2016.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Sautenet B, Cho Y, Gutman T, Rangan G, Ong A, Chapman AB, Ahn C, Coolican H, Tze-Wah Kao J, Fowler K, Gansevoort RT, Geneste C, Perrone RD, Harris T, Torres VE, Pei Y, Craig JC, Tong A; SONG-PKD Initiative. Range and variability of outcomes reported in randomized trials conducted in patients with polycystic kidney disease: a systematic review. Am J Kidney Dis 76: 213–223, 2020. doi: 10.1053/j.ajkd.2019.12.003. [DOI] [PubMed] [Google Scholar]
- 8. Lanktree MB, Chapman AB. New treatment paradigms for ADPKD: moving towards precision medicine. Nat Rev Nephrol 13: 750–768, 2017. doi: 10.1038/nrneph.2017.127. [DOI] [PubMed] [Google Scholar]
- 9. Schrier RW. ACE inhibitors, left ventricular mass and renal cyst growth in ADPKD. Pharmacol Res 114: 166–168, 2016. doi: 10.1016/j.phrs.2016.10.002. [DOI] [PubMed] [Google Scholar]
- 10. Grantham JJ, Geiser JL, Evan AP. Cyst formation and growth in autosomal dominant polycystic kidney disease. Kidney Int 31: 1145–1152, 1987. doi: 10.1038/ki.1987.121. [DOI] [PubMed] [Google Scholar]
- 11. Ye M, Grantham JJ. The secretion of fluid by renal cysts from patients with autosomal dominant polycystic kidney disease. N Engl J Med 329: 310–313, 1993. doi: 10.1056/NEJM199307293290503. [DOI] [PubMed] [Google Scholar]
- 12. Reif GA, Wallace DP. ADPKD cell proliferation and Cl-dependent fluid secretion. Methods Cell Biol 153: 69–92, 2019. doi: 10.1016/bs.mcb.2019.06.001. [DOI] [PubMed] [Google Scholar]
- 13. Yamaguchi T, Pelling JC, Ramaswamy NT, Eppler JW, Wallace DP, Nagao S, Rome LA, Sullivan LP, Grantham JJ. cAMP stimulates the in vitro proliferation of renal cyst epithelial cells by activating the extracellular signal-regulated kinase pathway. Kidney Int 57: 1460–1471, 2000. doi: 10.1046/j.1523-1755.2000.00991.x. [DOI] [PubMed] [Google Scholar]
- 14. Yamaguchi T, Nagao S, Wallace DP, Belibi FA, Cowley BD, Pelling JC, Grantham JJ. Cyclic AMP activates B-Raf and ERK in cyst epithelial cells from autosomal-dominant polycystic kidneys. Kidney Int 63: 1983–1994, 2003. doi: 10.1046/j.1523-1755.2003.00023.x. [DOI] [PubMed] [Google Scholar]
- 15. Belibi FA, Reif G, Wallace DP, Yamaguchi T, Olsen L, Li H, Helmkamp GM Jr, Grantham JJ. Cyclic AMP promotes growth and secretion in human polycystic kidney epithelial cells. Kidney Int 66: 964–973, 2004. doi: 10.1111/j.1523-1755.2004.00843.x. [DOI] [PubMed] [Google Scholar]
- 16. Wallace DP. Cyclic AMP-mediated cyst expansion. Biochim Biophys Acta 1812: 1291–1300, 2011. doi: 10.1016/j.bbadis.2010.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sussman CR, Wang X, Chebib FT, Torres VE. Modulation of polycystic kidney disease by G-protein coupled receptors and cyclic AMP signaling. Cell Signal 72: 109649, 2020. doi: 10.1016/j.cellsig.2020.109649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Rajagopal M, Wallace DP. Chloride secretion by renal collecting ducts. Curr Opin Nephrol Hypertens 24: 444–449, 2015. doi: 10.1097/MNH.0000000000000148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Yamaguchi T, Wallace DP, Magenheimer BS, Hempson SJ, Grantham JJ, Calvet JP. Calcium restriction allows cAMP activation of the B-Raf/ERK pathway, switching cells to a cAMP-dependent growth-stimulated phenotype. J Biol Chem 279: 40419–40430, 2004. doi: 10.1074/jbc.M405079200. [DOI] [PubMed] [Google Scholar]
- 20. Yamaguchi T, Hempson SJ, Reif GA, Hedge AM, Wallace DP. Calcium restores a normal proliferation phenotype in human polycystic kidney disease epithelial cells. J Am Soc Nephrol 17: 178–187, 2006. doi: 10.1681/ASN.2005060645. [DOI] [PubMed] [Google Scholar]
- 21. Liu Y, Dai B, Fu L, Jia J, Mei C. Rosiglitazone inhibits cell proliferation by inducing G1 cell cycle arrest and apoptosis in ADPKD cyst-lining epithelia cells. Basic Clin Pharmacol Toxicol 106: 523–530, 2010. doi: 10.1111/j.1742-7843.2010.00539.x. [DOI] [PubMed] [Google Scholar]
- 22. Buchholz B, Klanke B, Schley G, Bollag G, Tsai J, Kroening S, Yoshihara D, Wallace DP, Kraenzlin B, Gretz N, Hirth P, Eckardt KU, Bernhardt WM. The Raf kinase inhibitor PLX5568 slows cyst proliferation in rat polycystic kidney disease but promotes renal and hepatic fibrosis. Nephrol Dial Transplant 26: 3458–3465, 2011. doi: 10.1093/ndt/gfr432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Bukanov NO, Moreno SE, Natoli TA, Rogers KA, Smith LA, Ledbetter SR, Oumata N, Galons H, Meijer L, Ibraghimov-Beskrovnaya O. CDK inhibitors R-roscovitine and S-CR8 effectively block renal and hepatic cystogenesis in an orthologous model of ADPKD. Cell Cycle 11: 4040–4046, 2012. doi: 10.4161/cc.22375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Shillingford JM, Leamon CP, Vlahov IR, Weimbs T. Folate-conjugated rapamycin slows progression of polycystic kidney disease. J Am Soc Nephrol 23: 1674–1681, 2012. doi: 10.1681/ASN.2012040367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Caroli A, Perico N, Perna A, Antiga L, Brambilla P, Pisani A, Visciano B, Imbriaco M, Messa P, Cerutti R, Dugo M, Cancian L, Buongiorno E, De Pascalis A, Gaspari F, Carrara F, Rubis N, Prandini S, Remuzzi A, Remuzzi G, Ruggenenti P; ALADIN Study Group. Effect of long-acting somatostatin analogue on kidney and cyst growth in autosomal dominant polycystic kidney disease (ALADIN): a randomised, placebo-controlled, multicentre trial. Lancet 382: 1485–1495, 2013. doi: 10.1016/S0140-6736(13)61407-5. [DOI] [PubMed] [Google Scholar]
- 26. Rowe I, Chiaravalli M, Mannella V, Ulisse V, Quilici G, Pema M, Song XW, Xu H, Mari S, Qian F, Pei Y, Musco G, Boletta A. Defective glucose metabolism in polycystic kidney disease identifies a new therapeutic strategy. Nat Med 19: 488–493, 2013. doi: 10.1038/nm.3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Harris PC, Torres VE. Genetic mechanisms and signaling pathways in autosomal dominant polycystic kidney disease. J Clin Invest 124: 2315–2324, 2014. doi: 10.1172/JCI72272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. LaRiviere WB, Irazabal MV, Torres VE. Novel therapeutic approaches to autosomal dominant polycystic kidney disease. Transl Res 165: 488–498, 2015. doi: 10.1016/j.trsl.2014.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Ong ACM, Devuyst O, Knebelmann B, Walz G; ERA-EDTA Working Group for Inherited Diseases. Autosomal dominant polycystic kidney disease: the changing face of clinical management. Lancet 385: 1993–2002, 2015. doi: 10.1016/S0140-6736(15)60907-2. [DOI] [PubMed] [Google Scholar]
- 30. Hopp K, Hommerding CJ, Wang X, Ye H, Harris PC, Torres VE. Tolvaptan plus pasireotide shows enhanced efficacy in a PKD1 model. J Am Soc Nephrol 26: 39–47, 2015. doi: 10.1681/ASN.2013121312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Nikonova AS, Deneka AY, Eckman L, Kopp MC, Hensley HH, Egleston BL, Golemis EA. Opposing effects of inhibitors of Aurora-A and EGFR in autosomal-dominant polycystic kidney disease. Front Oncol 5: 228, 2015. doi: 10.3389/fonc.2015.00228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Hofherr A, Wagner CJ, Watnick T, Köttgen M. Targeted rescue of a polycystic kidney disease mutation by lysosomal inhibition. Kidney Int 89: 949–955, 2016. doi: 10.1016/j.kint.2015.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Hwang VJ, Zhou X, Chen X, Trott J, Abu Aboud O, Shim K, Dionne LK, Chmiel KJ, Senapedis W, Baloglu E, Mahjoub MR, Li X, Weiss RH. Anticystogenic activity of a small molecule PAK4 inhibitor may be a novel treatment for autosomal dominant polycystic kidney disease. Kidney Int 92: 922–933, 2017. doi: 10.1016/j.kint.2017.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Griffiths J, Mills MT, Ong AC. Long-acting somatostatin analogue treatments in autosomal dominant polycystic kidney disease and polycystic liver disease: a systematic review and meta-analysis. BMJ Open 10: e032620, 2020. doi: 10.1136/bmjopen-2019-032620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Torres VE, Chapman AB, Devuyst O, Gansevoort RT, Grantham JJ, Higashihara E, Perrone RD, Krasa HB, Ouyang J, Czerwiec FS, Investigators TT. Tolvaptan in patients with autosomal dominant polycystic kidney disease. N Engl J Med 367: 2407–2418, 2012. doi: 10.1056/NEJMoa1205511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Torres VE, Higashihara E, Devuyst O, Chapman AB, Gansevoort RT, Grantham JJ, Perrone RD, Ouyang J, Blais JD, Czerwiec FS; for the TEMPO 3:4 Trial Investigators. Effect of tolvaptan in autosomal dominant polycystic kidney disease by CKD stage: results from the TEMPO 3:4 trial. Clin J Am Soc Nephrol 11: 803–811, 2016. doi: 10.2215/CJN.06300615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Edwards ME, Chebib FT, Irazabal MV, Ofstie TG, Bungum LA, Metzger AJ, Senum SR, Hogan MC, El-Zoghby ZM, Kline TL, Harris PC, Czerwiec FS, Torres VE. Long-term administration of tolvaptan in autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol 13: 1153–1161, 2018. doi: 10.2215/CJN.01520218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Chebib FT, Perrone RD, Chapman AB, Dahl NK, Harris PC, Mrug M, Mustafa RA, Rastogi A, Watnick T, Yu ASL, Torres VE. A practical guide for treatment of rapidly progressive ADPKD with tolvaptan. J Am Soc Nephrol 29: 2458–2470, 2018. doi: 10.1681/ASN.2018060590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Blair HA. Tolvaptan: a review in autosomal dominant polycystic kidney disease. Drugs 79: 303–313, 2019. doi: 10.1007/s40265-019-1056-1. [DOI] [PubMed] [Google Scholar]
- 40. Nobakht N, Hanna RM, Al-Baghdadi M, Ameen KM, Arman F, Nobahkt E, Kamgar M, Rastogi A. Advances in autosomal dominant polycystic kidney disease: a clinical review. Kidney Med 2: 196–208, 2020. doi: 10.1016/j.xkme.2019.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Chang MY, Ong ACM. Targeting new cellular disease pathways in autosomal dominant polycystic kidney disease. Nephrol Dial Transplant 33: 1310–1316, 2018. doi: 10.1093/ndt/gfx262. [DOI] [PubMed] [Google Scholar]
- 42. Xue C, Dai B, Mei C. Long-term treatment with mammalian target of rapamycin inhibitor does not benefit patients with autosomal dominant polycystic kidney disease: a meta-analysis. Nephron Clin Pract 124: 10–16, 2013. doi: 10.1159/000354398. [DOI] [PubMed] [Google Scholar]
- 43. Wang CJ, Grantham JJ, Wetmore JB. The medicinal use of water in renal disease. Kidney Int 84: 45–53, 2013. doi: 10.1038/ki.2013.23. [DOI] [PubMed] [Google Scholar]
- 44. Torres VE, Bankir L, Grantham JJ. A case for water in the treatment of polycystic kidney disease. Clin J Am Soc Nephrol 4: 1140–1150, 2009. doi: 10.2215/CJN.00790209. [DOI] [PubMed] [Google Scholar]
- 45. Wong ATY, Mannix C, Grantham JJ, Allman-Farinelli M, Badve SV, Boudville N, Byth K, Chan J, Coulshed S, Edwards ME, Erickson BJ, Fernando M, Foster S, Haloob I, Harris DCH, Hawley CM, Hill J, Howard K, Howell M, Jiang SH, Johnson DW, Kline TL, Kumar K, Lee VW, Lonergan M, Mai J, McCloud P, Peduto A, Rangan A, Roger SD, Sud K, Torres V, Vilayur E, Rangan GK. Randomised controlled trial to determine the efficacy and safety of prescribed water intake to prevent kidney failure due to autosomal dominant polycystic kidney disease (PREVENT-ADPKD). BMJ Open 8: e018794, 2018. doi: 10.1136/bmjopen-2017-018794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Leone MG, Grippa E, Guidolin D, Tita B, Abdel-Haq H, Gatto MT, Bordi F, Cheng CY, Silvestrini B, Saso L. Effects of lonidamine on testicular and epididymal proteins in the rat. Reprod Toxicol 14: 257–263, 2000. doi: 10.1016/S0890-6238(00)00076-9. [DOI] [PubMed] [Google Scholar]
- 47. Nath K, Guo L, Nancolas B, Nelson DS, Shestov AA, Lee SC, Roman J, Zhou R, Leeper DB, Halestrap AP, Blair IA, Glickson JD. Mechanism of antineoplastic activity of lonidamine. Biochim Biophys Acta 1866: 151–162, 2016. doi: 10.1016/j.bbcan.2016.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Gong XD, Wong PY. Characterization of lonidamine and AF2785 blockade of the cyclic AMP-activated chloride current in rat epididymal cells. J Membr Biol 178: 225–233, 2000. doi: 10.1007/s002320010030. [DOI] [PubMed] [Google Scholar]
- 49. Gong XD, Wong YL, Leung GP, Cheng CY, Silvestrini B, Wong PY. Lonidamine and analogue AF2785 block the cyclic adenosine 3', 5'-monophosphate-activated chloride current and chloride secretion in the rat epididymis. Biol Reprod 63: 833–838, 2000. doi: 10.1095/biolreprod63.3.833. [DOI] [PubMed] [Google Scholar]
- 50. Gong X, Burbridge SM, Lewis AC, Wong PY, Linsdell P. Mechanism of lonidamine inhibition of the CFTR chloride channel. Br J Pharmacol 137: 928–936, 2002. doi: 10.1038/sj.bjp.0704932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Di Cosimo S, Ferretti G, Papaldo P, Carlini P, Fabi A, Cognetti F. Lonidamine: efficacy and safety in clinical trials for the treatment of solid tumors. Drugs Today (Barc) 39: 157–174, 2003. doi: 10.1358/dot.2003.39.3.799451. [DOI] [PubMed] [Google Scholar]
- 52. Tash JS, Chakrasali R, Jakkaraj SR, Hughes J, Smith SK, Hornbaker K, Heckert LL, Ozturk SB, Hadden MK, Kinzy TG, Blagg BS, Georg GI. Gamendazole, an orally active indazole carboxylic acid male contraceptive agent, targets HSP90AB1 (HSP90BETA) and EEF1A1 (eEF1A), and stimulates Il1a transcription in rat Sertoli cells. Biol Reprod 78: 1139–1152, 2008. doi: 10.1095/biolreprod.107.062679. [DOI] [PubMed] [Google Scholar]
- 53. Matts RL, Brandt GE, Lu Y, Dixit A, Mollapour M, Wang S, Donnelly AC, Neckers L, Verkhivker G, Blagg BS. A systematic protocol for the characterization of Hsp90 modulators. Bioorg Med Chem 19: 684–692, 2011. doi: 10.1016/j.bmc.2010.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Olesen SH, Ingles DJ, Zhu JY, Martin MP, Betzi S, Georg GI, Tash JS, Schönbrunn E. Stability of the human Hsp90-p50Cdc37 chaperone complex against nucleotides and Hsp90 inhibitors, and the influence of phosphorylation by casein kinase 2. Molecules 20: 1643–1660, 2015. doi: 10.3390/molecules20011643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Tash JS, Attardi B, Hild SA, Chakrasali R, Jakkaraj SR, Georg GI. A novel potent indazole carboxylic acid derivative blocks spermatogenesis and is contraceptive in rats after a single oral dose. Biol Reprod 78: 1127–1138, 2008. doi: 10.1095/biolreprod.106.057810. [DOI] [PubMed] [Google Scholar]
- 56. Taipale M, Jarosz DF, Lindquist S. HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat Rev Mol Cell Biol 11: 515–528, 2010. doi: 10.1038/nrm2918. [DOI] [PubMed] [Google Scholar]
- 57. Li J, Buchner J. Structure, function and regulation of the hsp90 machinery. Biomed J 36: 106–117, 2013. doi: 10.4103/2319-4170.113230. [DOI] [PubMed] [Google Scholar]
- 58. Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev Cancer 5: 761–772, 2005. doi: 10.1038/nrc1716. [DOI] [PubMed] [Google Scholar]
- 59. Taldone T, Gozman A, Maharaj R, Chiosis G. Targeting Hsp90: small-molecule inhibitors and their clinical development. Curr Opin Pharmacol 8: 370–374, 2008. doi: 10.1016/j.coph.2008.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Pearl LH, Prodromou C, Workman P. The Hsp90 molecular chaperone: an open and shut case for treatment. Biochem J 410: 439–453, 2008. doi: 10.1042/BJ20071640. [DOI] [PubMed] [Google Scholar]
- 61. Trepel J, Mollapour M, Giaccone G, Neckers L. Targeting the dynamic HSP90 complex in cancer. Nat Rev Cancer 10: 537–549, 2010. doi: 10.1038/nrc2887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Jafari A, Rezaei-Tavirani M, Farhadihosseinabadi B, Taranejoo S, Zali H. HSP90 and co-chaperones: impact on tumor progression and prospects for molecular-targeted cancer therapy. Cancer Invest 38: 310–328, 2020. doi: 10.1080/07357907.2020.1752227. [DOI] [PubMed] [Google Scholar]
- 63. Costa T, Raghavendra NM, Penido C. Natural heat shock protein 90 inhibitors in cancer and inflammation. Eur J Med Chem 189: 112063, 2020. doi: 10.1016/j.ejmech.2020.112063. [DOI] [PubMed] [Google Scholar]
- 64. Wilson SJ, Amsler K, Hyink DP, Li X, Lu W, Zhou J, Burrow CR, Wilson PD. Inhibition of HER-2(neu/ErbB2) restores normal function and structure to polycystic kidney disease (PKD) epithelia. Biochim Biophys Acta 1762: 647–655, 2006. doi: 10.1016/j.bbadis.2006.04.006. [DOI] [PubMed] [Google Scholar]
- 65. Nakanishi K, Sweeney W Jr, Avner ED. Segment-specific c-ErbB2 expression in human autosomal recessive polycystic kidney disease. J Am Soc Nephrol 12: 379–384, 2001. doi: 10.1681/ASN.V122379. [DOI] [PubMed] [Google Scholar]
- 66. Yang B, Sonawane ND, Zhao D, Somlo S, Verkman AS. Small-molecule CFTR inhibitors slow cyst growth in polycystic kidney disease. J Am Soc Nephrol 19: 1300–1310, 2008. doi: 10.1681/ASN.2007070828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Magenheimer BS, St John PL, Isom KS, Abrahamson DR, De Lisle RC, Wallace DP, Maser RL, Grantham JJ, Calvet JP. Early embryonic renal tubules of wild-type and polycystic kidney disease kidneys respond to cAMP stimulation with cystic fibrosis transmembrane conductance regulator/Na+, K+, 2Cl- co-transporter-dependent cystic dilation. J Am Soc Nephrol 17: 3424–3437, 2006. doi: 10.1681/ASN.2006030295. [DOI] [PubMed] [Google Scholar]
- 68. Natoli TA, Smith LA, Rogers KA, Wang B, Komarnitsky S, Budman Y, Belenky A, Bukanov NO, Dackowski WR, Husson H, Russo RJ, Shayman JA, Ledbetter SR, Leonard JP, Ibraghimov-Beskrovnaya O. Inhibition of glucosylceramide accumulation results in effective blockade of polycystic kidney disease in mouse models. Nat Med 16: 788–792, 2010. doi: 10.1038/nm.2171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Boletta A. Emerging evidence of a link between the polycystins and the mTOR pathways. Pathogenetics 2: 6, 2009. doi: 10.1186/1755-8417-2-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Bukanov NO, Smith LA, Klinger KW, Ledbetter SR, Ibraghimov-Beskrovnaya O. Long-lasting arrest of murine polycystic kidney disease with CDK inhibitor roscovitine. Nature 444: 949–952, 2006. doi: 10.1038/nature05348. [DOI] [PubMed] [Google Scholar]
- 71. Yamaguchi T, Reif GA, Calvet JP, Wallace DP. Sorafenib inhibits cAMP-dependent ERK activation, cell proliferation, and in vitro cyst growth of human ADPKD cyst epithelial cells. Am J Physiol Renal Physiol 299: F944–F951, 2010. doi: 10.1152/ajprenal.00387.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Park EY, Sung YH, Yang MH, Noh JY, Park SY, Lee TY, Yook YJ, Yoo KH, Roh KJ, Kim I, Hwang YH, Oh GT, Seong JK, Ahn C, Lee HW, Park JH. Cyst formation in kidney via B-Raf signaling in the PKD2 transgenic mice. J Biol Chem 284: 7214–7222, 2009. doi: 10.1074/jbc.M805890200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Shibazaki S, Yu Z, Nishio S, Tian X, Thomson RB, Mitobe M, Louvi A, Velazquez H, Ishibe S, Cantley LG, Igarashi P, Somlo S. Cyst formation and activation of the extracellular regulated kinase pathway after kidney specific inactivation of Pkd1. Hum Mol Genet 17: 1505–1516, 2008. doi: 10.1093/hmg/ddn039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Parker E, Newby LJ, Sharpe CC, Rossetti S, Streets AJ, Harris PC, O'Hare MJ, Ong AC. Hyperproliferation of PKD1 cystic cells is induced by insulin-like growth factor-1 activation of the Ras/Raf signalling system. Kidney Int 72: 157–165, 2007. doi: 10.1038/sj.ki.5002229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Smithline ZB, Nikonova AS, Hensley HH, Cai KQ, Egleston BL, Proia DA, Seeger-Nukpezah T, Golemis EA. Inhibiting heat shock protein 90 (HSP90) limits the formation of liver cysts induced by conditional deletion of Pkd1 in mice. PLoS One 9: e114403, 2014. doi: 10.1371/journal.pone.0114403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Seeger-Nukpezah T, Proia DA, Egleston BL, Nikonova AS, Kent T, Cai KQ, Hensley HH, Ying W, Chimmanamada D, Serebriiskii IG, Golemis EA. Inhibiting the HSP90 chaperone slows cyst growth in a mouse model of autosomal dominant polycystic kidney disease. Proc Natl Acad Sci USA 110: 12786–12791, 2013. doi: 10.1073/pnas.1301904110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Nikonova AS, Deneka AY, Kiseleva AA, Korobeynikov V, Gaponova A, Serebriiskii IG, Kopp MC, Hensley HH, Seeger-Nukpezah TN, Somlo S, Proia DA, Golemis EA. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD). FASEB J 32: 2735–2746, 2018. doi: 10.1096/fj.201700909R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Wallace DP, Grantham JJ, Sullivan LP. Chloride and fluid secretion by cultured human polycystic kidney cells. Kidney Int 50: 1327–1336, 1996. doi: 10.1038/ki.1996.445. [DOI] [PubMed] [Google Scholar]
- 79. Wallace DP, Reif GA. Generation of primary cells from ADPKD and normal human kidneys. Methods Cell Biol 153: 1–23, 2019. doi: 10.1016/bs.mcb.2019.05.005. [DOI] [PubMed] [Google Scholar]
- 80. Stoos BA, Náray-Fejes-Tóth A, Carretero OA, Ito S, Fejes-Tóth G. Characterization of a mouse cortical collecting duct cell line. Kidney Int 39: 1168–1175, 1991. doi: 10.1038/ki.1991.148. [DOI] [PubMed] [Google Scholar]
- 81. Herron BJ, Lu W, Rao C, Liu S, Peters H, Bronson RT, Justice MJ, McDonald JD, Beier DR. Efficient generation and mapping of recessive developmental mutations using ENU mutagenesis. Nat Genet 30: 185–189, 2002. doi: 10.1038/ng812. [DOI] [PubMed] [Google Scholar]
- 82. Avner ED, Ellis D, Temple T, Jaffe R. Metanephric development in serum-free organ culture. In Vitro 18: 675–682, 1982. doi: 10.1007/BF02796422. [DOI] [PubMed] [Google Scholar]
- 83. Welham SJ, Woolf AS. Organ culture of intact metanephric kidneys. Methods Mol Med 86: 169–177, 2003. doi: 10.1385/1-59259-392-5:169. [DOI] [PubMed] [Google Scholar]
- 84. Maser RL, Magenheimer BS, Calvet JP. Metanephric organ culture. Methods Cell Biol 153: 169–183, 2019. doi: 10.1016/bs.mcb.2019.04.018. [DOI] [PubMed] [Google Scholar]
- 85. Akner G, Mossberg K, Sundqvist KG, Gustafsson JA, Wikstrom AC. Evidence for reversible, non-microtubule and non-microfilament-dependent nuclear translocation of hsp90 after heat shock in human fibroblasts. Eur J Cell Biol 58: 356–364, 1992. [PubMed] [Google Scholar]
- 86. Sidera K, Patsavoudi E. HSP90 inhibitors: current development and potential in cancer therapy. Recent Pat Anticancer Drug Discov 9: 1–20, 2014. doi: 10.2174/15748928113089990031. [DOI] [PubMed] [Google Scholar]
- 87. Matei D, Satpathy M, Cao L, Lai YC, Nakshatri H, Donner DB. The platelet-derived growth factor receptor alpha is destabilized by geldanamycins in cancer cells. J Biol Chem 282: 445–453, 2007. doi: 10.1074/jbc.M607012200. [DOI] [PubMed] [Google Scholar]
- 88. Loo MA, Jensen TJ, Cui L, Hou Y, Chang XB, Riordan JR. Perturbation of Hsp90 interaction with nascent CFTR prevents its maturation and accelerates its degradation by the proteasome. EMBO J 17: 6879–6887, 1998. doi: 10.1093/emboj/17.23.6879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Joly D, Morel V, Hummel A, Ruello A, Nusbaum P, Patey N, Noel LH, Rousselle P, Knebelmann B. β4 integrin and laminin 5 are aberrantly expressed in polycystic kidney disease: role in increased cell adhesion and migration. Am J Pathol 163: 1791–1800, 2003. doi: 10.1016/S0002-9440(10)63539-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Murray JW, Edmonds BT, Liu G, Condeelis J. Bundling of actin filaments by elongation factor 1 alpha inhibits polymerization at filament ends. J Cell Biol 135: 1309–1321, 1996. doi: 10.1083/jcb.135.5.1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Gross SR, Kinzy TG. Translation elongation factor 1A is essential for regulation of the actin cytoskeleton and cell morphology. Nat Struct Mol Biol 12: 772–778, 2005. doi: 10.1038/nsmb979. [DOI] [PubMed] [Google Scholar]
- 92. Kamal A, Thao L, Sensintaffar J, Zhang L, Boehm MF, Fritz LC, Burrows FJ. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature 425: 407–410, 2003. doi: 10.1038/nature01913. [DOI] [PubMed] [Google Scholar]
- 93. Graner MW. HSP90 and immune modulation in cancer. Adv Cancer Res 129: 191–224, 2016. doi: 10.1016/bs.acr.2015.10.001. [DOI] [PubMed] [Google Scholar]
- 94. Solárová Z, Mojžiš J, Solár P. Hsp90 inhibitor as a sensitizer of cancer cells to different therapies (review). Int J Oncol 46: 907–926, 2015. doi: 10.3892/ijo.2014.2791. [DOI] [PubMed] [Google Scholar]
- 95. Seeger-Nukpezah T, Geynisman DM, Nikonova AS, Benzing T, Golemis EA. The hallmarks of cancer: relevance to the pathogenesis of polycystic kidney disease. Nat Rev Nephrol 11: 515–534, 2015. doi: 10.1038/nrneph.2015.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Grantham JJ. Polycystic kidney disease: neoplasia in disguise. Am J Kidney Dis 15: 110–116, 1990. doi: 10.1016/S0272-6386(12)80507-5. [DOI] [PubMed] [Google Scholar]
- 97. Hanaoka K, Devuyst O, Schwiebert EM, Wilson PD, Guggino WB. A role for CFTR in human autosomal dominant polycystic kidney disease. Am J Physiol Cell Physiol 270: C389–C399, 1996. doi: 10.1152/ajpcell.1996.270.1.C389. [DOI] [PubMed] [Google Scholar]
- 98. Davidow CJ, Maser RL, Rome LA, Calvet JP, Grantham JJ. The cystic fibrosis transmembrane conductance regulator mediates transepithelial fluid secretion by human autosomal dominant polycystic kidney disease epithelium in vitro. Kidney Int 50: 208–218, 1996. doi: 10.1038/ki.1996.304. [DOI] [PubMed] [Google Scholar]
- 99. Cowley BD Jr, Smardo FL Jr, Grantham JJ, Calvet JP. Elevated c-myc protooncogene expression in autosomal recessive polycystic kidney disease. Proc Natl Acad Sci USA 84: 8394–8398, 1987. [Erratum in Proc Natl Acad Sci USA 85: 2578, 1988]. doi: 10.1073/pnas.84.23.8394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. MacKay K, Striker LJ, Pinkert CA, Brinster RL, Striker GE. Glomerulosclerosis and renal cysts in mice transgenic for the early region of SV40. Kidney Int 32: 827–837, 1987. doi: 10.1038/ki.1987.283. [DOI] [PubMed] [Google Scholar]
- 101. Trudel M, D'Agati V, Costantini F. C-myc as an inducer of polycystic kidney disease in transgenic mice. Kidney Int 39: 665–671, 1991. doi: 10.1038/ki.1991.80. [DOI] [PubMed] [Google Scholar]
- 102. Schaffner DL, Barrios R, Massey C, Bañez EI, Ou CN, Rajagopalan S, Aguilar-Cordova E, Lebovitz RM, Overbeek PA, Lieberman MW. Targeting of the rasT24 oncogene to the proximal convoluted tubules in transgenic mice results in hyperplasia and polycystic kidneys. Am J Pathol 142: 1051–1060, 1993. [PMC free article] [PubMed] [Google Scholar]
- 103. Fukuyo Y, Hunt CR, Horikoshi N. Geldanamycin and its anti-cancer activities. Cancer Lett 290: 24–35, 2010. doi: 10.1016/j.canlet.2009.07.010. [DOI] [PubMed] [Google Scholar]
- 104. Montagut C, Settleman J. Targeting the RAF-MEK-ERK pathway in cancer therapy. Cancer Lett 283: 125–134, 2009. doi: 10.1016/j.canlet.2009.01.022. [DOI] [PubMed] [Google Scholar]
- 105. Whittaker S, Marais R, Zhu AX. The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene 29: 4989–5005, 2010. doi: 10.1038/onc.2010.236. [DOI] [PubMed] [Google Scholar]
- 106. Qian CN, Knol J, Igarashi P, Lin F, Zylstra U, Teh BT, Williams BO. Cystic renal neoplasia following conditional inactivation of apc in mouse renal tubular epithelium. J Biol Chem 280: 3938–3945, 2005. doi: 10.1074/jbc.M410697200. [DOI] [PubMed] [Google Scholar]
- 107. Ikeda M, Fong P, Cheng J, Boletta A, Qian F, Zhang XM, Cai H, Germino GG, Guggino WB. A regulatory role of polycystin-1 on cystic fibrosis transmembrane conductance regulator plasma membrane expression. Cell Physiol Biochem 18: 9–20, 2006. doi: 10.1159/000095133. [DOI] [PubMed] [Google Scholar]
- 108. Ditonno P, Battaglia M, Selvaggio O, Garofalo L, Lorusso V, Selvaggi FP. Clinical evidence supporting the role of lonidamine for the treatment of BPH. Rev Urol 7, Suppl 7: S27–S33, 2005. [PMC free article] [PubMed] [Google Scholar]
- 109. Brawer MK. Lonidamine: basic science and rationale for treatment of prostatic proliferative disorders. Rev Urol 7, Suppl 7: S21–S26, 2005. [PMC free article] [PubMed] [Google Scholar]
- 110. Fanciulli M, Valentini A, Bruno T, Citro G, Zupi G, Floridi A. Effect of the antitumor drug lonidamine on glucose metabolism of adriamycin-sensitive and -resistant human breast cancer cells. Oncol Res 8: 111–120, 1996. [PubMed] [Google Scholar]
- 111. Paggi MG, Fanciulli M, Perrotti N, Floridi A, Zeuli M, Silvestrini B, Caputo A. The role of mitochondrial hexokinase in neoplastic phenotype and its sensitivity to lonidamine. Ann NY Acad Sci 551: 358–360, 1988. doi: 10.1111/j.1749-6632.1988.tb22362.x. [DOI] [PubMed] [Google Scholar]
- 112. Silvestrini B, Caputo A. Lonidamine: a new pharmacological approach to the study and control of spermatogenesis and tumors. Chemotherapy 27, Suppl 2: 1–120, 1981. doi: 10.1159/000238037. [DOI] [PubMed] [Google Scholar]
- 113. Shillingford JM, Murcia NS, Larson CH, Low SH, Hedgepeth R, Brown N, Flask CA, Novick AC, Goldfarb DA, Kramer-Zucker A, Walz G, Piontek KB, Germino GG, Weimbs T. The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease. Proc Natl Acad Sci USA 103: 5466–5471, 2006. doi: 10.1073/pnas.0509694103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Sullivan LP, Wallace DP, Grantham JJ. Chloride and fluid secretion in polycystic kidney disease. J Am Soc Nephrol 9: 903–916, 1998. doi: 10.1681/ASN.V95903. [DOI] [PubMed] [Google Scholar]
- 115. Del Bufalo D, Trisciuoglio D, Scarsella M, D'Amati G, Candiloro A, Iervolino A, Leonetti C, Zupi G. Lonidamine causes inhibition of angiogenesis-related endothelial cell functions. Neoplasia 6: 513–522, 2004. doi: 10.1593/neo.04133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Malorni W, Meschini S, Matarrese P, Arancia G. The cytoskeleton as a subcellular target of the antineoplastic drug lonidamine. Anticancer Res 12: 2037–2045, 1992. [PubMed] [Google Scholar]
- 117. Demma M, Warren V, Hock R, Dharmawardhane S, Condeelis J. Isolation of an abundant 50,000-dalton actin filament bundling protein from Dictyostelium amoebae. J Biol Chem 265: 2286–2291, 1990. doi: 10.1016/S0021-9258(19)39973-9. [DOI] [PubMed] [Google Scholar]
- 118. Whitesell L, Bagatell R, Falsey R. The stress response: implications for the clinical development of hsp90 inhibitors. Curr Cancer Drug Targets 3: 349–358, 2003. doi: 10.2174/1568009033481787. [DOI] [PubMed] [Google Scholar]
- 119. Guo F, Rocha K, Bali P, Pranpat M, Fiskus W, Boyapalle S, Kumaraswamy S, Balasis M, Greedy B, Armitage ES, Lawrence N, Bhalla K. Abrogation of heat shock protein 70 induction as a strategy to increase antileukemia activity of heat shock protein 90 inhibitor 17-allylamino-demethoxy geldanamycin. Cancer Res 65: 10536–10544, 2005. doi: 10.1158/0008-5472.CAN-05-1799. [DOI] [PubMed] [Google Scholar]
- 120. Schmitt E, Gehrmann M, Brunet M, Multhoff G, Garrido C. Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy. J Leukoc Biol 81: 15–27, 2007. doi: 10.1189/jlb.0306167. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental Figs. S1−S7: https://doi.org/10.6084/m9.figshare.20371803.


